Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 45
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
J Immunol ; 201(10): 2910-2922, 2018 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-30291167

RESUMEN

Helminths stimulate the secretion of Th2 cytokines, like IL-4, and suppress lethal graft-versus-host disease (GVHD) after bone marrow transplantation. This suppression depends on the production of immune-modulatory TGF-ß and is associated with TGF-ß-dependent in vivo expansion of Foxp3+ regulatory T cells (Treg). In vivo expansion of Tregs is under investigation for its potential as a therapy for GVHD. Nonetheless, the mechanism of induced and TGF-ß-dependent in vivo expansion of Tregs, in a Th2 polarized environment after helminth infection, is unknown. In this study, we show that helminth-induced IL-4 production by host cells is critical to the induction and maintenance of TGF-ß secretion, TGF-ß-dependent expansion of Foxp3+ Tregs, and the suppression of GVHD. In mice with GVHD, the expanding donor Tregs express the Th2-driving transcription factor, GATA3, which is required for helminth-induced production of IL-4 and TGF-ß. In contrast, TGF-ß is not necessary for GATA3 expression by Foxp3+ Tregs or by Foxp3- CD4 T cells. Various cell types of innate or adaptive immune compartments produce high quantities of IL-4 after helminth infection. As a result, IL-4-mediated suppression of GVHD does not require invariant NKT cells of the host, a cell type known to produce IL-4 and suppress GVHD in other models. Thus, TGF-ß generation, in a manner dependent on IL-4 secretion by host cells and GATA3 expression, constitutes a critical effector arm of helminthic immune modulation that promotes the in vivo expansion of Tregs and suppresses GVHD.


Asunto(s)
Enfermedad Injerto contra Huésped/inmunología , Interleucina-4/biosíntesis , Infecciones por Strongylida/inmunología , Linfocitos T Reguladores/inmunología , Factor de Crecimiento Transformador beta/biosíntesis , Animales , Trasplante de Médula Ósea , Linfocitos T CD4-Positivos/inmunología , Factor de Transcripción GATA3/inmunología , Factor de Transcripción GATA3/metabolismo , Interleucina-4/inmunología , Activación de Linfocitos/inmunología , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Nematospiroides dubius , Factor de Crecimiento Transformador beta/inmunología
2.
J Biol Chem ; 292(34): 14066-14079, 2017 08 25.
Artículo en Inglés | MEDLINE | ID: mdl-28360105

RESUMEN

Type 1 diabetes (T1D) can be managed by transplanting either the whole pancreas or isolated pancreatic islets. However, cadaveric pancreas is scarcely available for clinical use, limiting this approach. As such, there is a great need to identify alternative sources of clinically usable pancreatic tissues. Here, we used induced pluripotent stem (iPS) cells derived from patients with T1D to generate glucose-responsive, insulin-producing cells (IPCs) via 3D culture. Initially, T1D iPS cells were resistant to differentiation, but transient demethylation treatment significantly enhanced IPC yield. The cells responded to high-glucose stimulation by secreting insulin in vitro The shape, size, and number of their granules, as observed by transmission electron microscopy, were identical to those found in cadaveric ß cells. When the IPCs were transplanted into immunodeficient mice that had developed streptozotocin-induced diabetes, they promoted a dramatic decrease in hyperglycemia, causing the mice to become normoglycemic within 28 days. None of the mice died or developed teratomas. Because the cells are derived from "self," immunosuppression is not required, providing a much safer and reliable treatment option for T1D patients. Moreover, these cells can be used for drug screening, thereby accelerating drug discovery. In conclusion, our approach eliminates the need for cadaveric pancreatic tissue.


Asunto(s)
Metilación de ADN/efectos de los fármacos , Metilasas de Modificación del ADN/antagonistas & inhibidores , Diabetes Mellitus Tipo 1/metabolismo , Células Madre Pluripotentes Inducidas/efectos de los fármacos , Células Secretoras de Insulina/metabolismo , Insulina/metabolismo , Organoides/metabolismo , Animales , Azacitidina/análogos & derivados , Azacitidina/farmacología , Cadáver , Diferenciación Celular/efectos de los fármacos , Células Cultivadas , Metilasas de Modificación del ADN/metabolismo , Decitabina , Diabetes Mellitus Experimental/sangre , Diabetes Mellitus Experimental/inmunología , Diabetes Mellitus Experimental/cirugía , Diabetes Mellitus Tipo 1/patología , Diabetes Mellitus Tipo 1/cirugía , Inhibidores Enzimáticos/farmacología , Humanos , Hiperglucemia/prevención & control , Células Madre Pluripotentes Inducidas/metabolismo , Células Madre Pluripotentes Inducidas/patología , Células Madre Pluripotentes Inducidas/ultraestructura , Insulina/biosíntesis , Secreción de Insulina , Células Secretoras de Insulina/trasplante , Células Secretoras de Insulina/ultraestructura , Ratones Noqueados , Microscopía Electrónica de Transmisión , Organoides/trasplante , Organoides/ultraestructura , Vesículas Secretoras/metabolismo , Vesículas Secretoras/ultraestructura , Andamios del Tejido , Trasplante Heterólogo/efectos adversos , Trasplante Heterotópico/efectos adversos
3.
Immunology ; 151(2): 191-197, 2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-28185259

RESUMEN

The safety of induced pluripotent stem cells (iPSCs) in autologous recipients has been questioned after iPSCs, but not embryonic stem cells (ESCs), were reported to be rejected in syngeneic mice. This important topic has remained controversial because there has not been a mechanistic explanation for this phenomenon. Here, we hypothesize that iPSCs, but not ESCs, readily differentiate into gamete-forming cells that express meiotic antigens normally found in immune-privileged gonads. Because peripheral blood T cells are not tolerized to these antigens in the thymus, gamete-associated-proteins (GAPs) sensitize T cells leading to rejection. Here, we provide evidence that GAPs expressed in iPSC teratomas, but not in ESC teratomas, are responsible for the immunological rejection of iPSCs. Furthermore, silencing the expression of Stra8, 'the master regulator of meiosis', in iPSCs, using short hairpin RNA led to significant abrogation of the rejection of iPSCs, supporting our central hypothesis that GAPs expressed after initiation of meiosis in iPSCs were responsible for rejection. In contrast to iPSCs, iPSC-derivatives, such as haematopoietic progenitor cells, are able to engraft long-term into syngeneic recipients because they no longer express GAPs. Our findings, for the first time, provide a unifying explanation of why iPSCs, but not ESCs, are rejected in syngeneic recipients, ending the current controversy on the safety of iPSCs and their derivatives.


Asunto(s)
Rechazo de Injerto/inmunología , Células Madre Pluripotentes Inducidas/inmunología , Células Madre Pluripotentes Inducidas/trasplante , Proteínas de la Membrana/inmunología , Animales , Línea Celular , Células Madre Pluripotentes Inducidas/metabolismo , Ratones , Linfocitos T/inmunología , Trasplante Isogénico
4.
Blood ; 121(26): 5167-75, 2013 Jun 27.
Artículo en Inglés | MEDLINE | ID: mdl-23687092

RESUMEN

Human induced pluripotent stem cells (iPSCs) have emerged as an alternative source of pluripotent stem cells that can be used for tissue regeneration in place of the controversial human embryonic stem cells. However, immunologic knowledge about iPSC derivatives remains enigmatic. Here, we characterized human iPS-derived CD34(+) hematopoietic progenitor cells (HPCs). These HPCs poorly express major histocompatibility complex (MHC) I antigens and are MHC-II negative. Interestingly, they moderately express nonclassical HLA-G and HLA-E molecules. Consequently, alloreactive HLA-A2-specific cytotoxic T cells failed to recognize HLA-A2-expressing HPCs but became anergic. Subsequent upregulation of MHC-I using interferon-γ stimulation and provision of CD28 cosignaling led to T-cell activation, confirming that poor delivery of signals 1 and 2 by the HPCs mediated T-cell anergy. These data indicate for the first time that HPCs induce T-cell anergy, a unique characteristic of iPSC-derived cells that confers immunologic advantage for allogenic transplantation. Although iPSCs are ideal for patient-tailored treatments with the anticipation that no immunosuppression will be required, in cases of gene defects, their derivatives could be used to treat diseases in nonhistocompatible recipients.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Fibroblastos/inmunología , Antígenos HLA/inmunología , Células Madre Hematopoyéticas/inmunología , Células Madre Pluripotentes Inducidas/inmunología , Linfocitos T Citotóxicos/inmunología , Células Cultivadas , Anergia Clonal , Células Madre Embrionarias/citología , Células Madre Embrionarias/efectos de los fármacos , Células Madre Embrionarias/inmunología , Fibroblastos/citología , Fibroblastos/efectos de los fármacos , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/efectos de los fármacos , Humanos , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/efectos de los fármacos , Interferón gamma/farmacología , Activación de Linfocitos/efectos de los fármacos
5.
Immunology ; 142(3): 421-30, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24527810

RESUMEN

Murine embryonic stem (ES) cell-derived haematopoietic progenitor cells (HPCs) engraft and populate lymphoid organs. In vivo, HPCs engraft across MHC barriers protecting donor-type allografts from rejection. However, the underlying phenomenon remains elusive. Here, we sought to determine the mechanism by which ES cell-derived HPCs regulate alloreactive T cells. We used the 2C mouse, which expresses a transgenic T-cell receptor against H2-L(d) to determine whether HPCs are deleted by cytotoxic T lymphocytes (CTLs). Previously, we reported that HPCs express MHC class I antigens poorly and do not express class II antigens. In vitro stimulated 2C CTLs failed to lyse H2-L(d) HPCs in a standard 4-hr (51) chromium release assay. Similarly, when the HPCs were tested in an ELISPOT assay measuring the release of interferon-γ by CTLs, HPCs failed to induce CTL degranulation. In addition, mice that were injected with HPCs showed a marked decrease in T-cell responses to alloantigen and CD3 stimulation, but showed a normal response to PMA/ionomycin, suggesting that HPCs impaired T-cell signalling through the T-cell receptor/CD3 complex. Here, we show that HPCs secrete arginase, an enzyme that scavenges l-arginine, leading to metabolites that down-regulate CD3 ζ chain. Indeed an arginase inhibitor partially restored expression of the CD3 ζ chain, implicating arginase 1 in the down-regulation of T cells. This previously unrecognized property of ES cell-derived HPCs could positively enhance the engraftment of ES cell-derived HPCs across MHC barriers by preventing rejection.


Asunto(s)
Complejo CD3/inmunología , Regulación hacia Abajo , Células Madre Embrionarias/citología , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/inmunología , Linfocitos T Reguladores/inmunología , Animales , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Endogámicos MRL lpr
6.
Blood ; 119(19): e139-47, 2012 May 10.
Artículo en Inglés | MEDLINE | ID: mdl-22438249

RESUMEN

Efficient in vitro generation of hematopoietic stem cells (HSCs) from embryonic stem cells (ESCs) holds great promise for cell-based therapies to treat hematologic diseases. To date, HoxB4 remains the most effective transcription factor (TF) the overexpression of which in ESCs confers long-term repopulating ability to ESC-derived HSCs. Despite its importance, the components and dynamics of the HoxB4 transcriptional regulatory network is poorly understood, hindering efforts to develop more efficient protocols for in vitro derivation of HSCs. In the present study, we performed global gene-expression profiling and ChIP coupled with deep sequencing at 4 stages of the HoxB4-mediated ESC differentiation toward HSCs. Joint analyses of ChIP/deep sequencing and gene-expression profiling unveiled several global features of the HoxB4 regulatory network. First, it is highly dynamic and gradually expands during the differentiation process. Second, HoxB4 functions as a master regulator of hematopoiesis by regulating multiple hematopoietic TFs and chromatin-modification enzymes. Third, HoxB4 acts in different combinations with 4 other hematopoietic TFs (Fli1, Meis1, Runx1, and Scl) to regulate distinct sets of pathways. Finally, the results of our study suggest that down-regulation of mitochondria and lysosomal genes by HoxB4 plays a role in the impaired lymphoid lineage development from ESC-derived HSCs.


Asunto(s)
Células Sanguíneas/fisiología , Diferenciación Celular/genética , Células Madre Embrionarias/fisiología , Redes Reguladoras de Genes/genética , Hematopoyesis/genética , Proteínas de Homeodominio/fisiología , Factores de Transcripción/fisiología , Animales , Células Sanguíneas/metabolismo , Diferenciación Celular/fisiología , Linaje de la Célula/genética , Linaje de la Célula/fisiología , Células Cultivadas , Células Madre Embrionarias/metabolismo , Perfilación de la Expresión Génica , Regulación del Desarrollo de la Expresión Génica , Hematopoyesis/fisiología , Células Madre Hematopoyéticas/metabolismo , Células Madre Hematopoyéticas/fisiología , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Humanos , Linfocitos/metabolismo , Linfocitos/fisiología , Ratones , Análisis por Micromatrices , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
7.
Curr Opin Organ Transplant ; 19(6): 598-602, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25333832

RESUMEN

PURPOSE OF REVIEW: Embryonic stem cells and induced pluripotent stem cells are pluripotent and therefore capable of differentiating into different cell types and tissues. However, their clinical potential, so far, has not been sufficiently probed. The major obstacle is the lack of protocols that allow efficient derivation of clinical grade cells or tissues. This review will address these questions and discuss the current state of the field. RECENT FINDINGS: I will address some of the ongoing clinical trials using stem cell-derived retinal pigment epithelial cells, cardiomyocytes, neurons and attempts to establish insulin-producing cells for the treatment of type 1 diabetes. SUMMARY: Are we there yet? The answer is clearly no. Progress in the different organs and tissues that are being generated is quite variable. Clearly, there has been more success in the derivation of retinal pigment epithelial cells, neuronal cells and cardiomyocytes than in any other tissues or organs. The derivation of insulin-producing cells and that of definitive hematopoietic progenitor cells in humans remains a challenge. Having said that the progress already made with other tissues is an encouraging sign that we may eventually see progress across the board.


Asunto(s)
Células Madre Embrionarias/inmunología , Células Madre Pluripotentes Inducidas/inmunología , Animales , Células Epiteliales/inmunología , Humanos , Miocitos Cardíacos/inmunología , Neuronas/inmunología , Trasplante de Células Madre
8.
FASEB J ; 24(2): 364-73, 2010 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19762558

RESUMEN

Bone marrow transplantation is a curative treatment for many diseases, including leukemia, autoimmune diseases, and a number of immunodeficiencies. Recently, it was claimed that bone marrow cells transdifferentiate, a much desired property as bone marrow cells are abundant and therefore could be used in regenerative medicine to treat incurable chronic diseases. Using a Cre/loxP system, we studied cell fusion after bone marrow transplantation. Fused cells were chiefly Gr-1(+), a myeloid cell marker, and found predominantly in the bone marrow; in parenchymal tissues. Surprisingly, fused cells were most abundant in the kidney, Peyer's patches, and cardiac tissue. In contrast, after cell fusion with embryonic stem cells, bone marrow cells were reprogrammed into new tetraploid pluripotent stem cells that successfully differentiated into beating cardiomyocytes. Together, these data suggest that cell fusion is ubiquitous after cellular transplants and that the subsequent sharing of genetic material between the fusion partners affects cellular survival and function. Fusion between tumor cells and bone marrow cells could have consequences for tumor malignancy.


Asunto(s)
Células de la Médula Ósea/fisiología , Fusión Celular , Células Madre Embrionarias/fisiología , Células Madre Pluripotentes/fisiología , Animales , Trasplante de Médula Ósea , Diferenciación Celular/fisiología , Reprogramación Celular/fisiología , Técnicas de Cocultivo , Integrasas/metabolismo , Ratones , Ratones Transgénicos , Miocitos Cardíacos/fisiología
9.
J Immunol ; 183(9): 5449-57, 2009 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-19828634

RESUMEN

Embryonic stem (ES) cells are a novel source of cells, especially hematopoietic progenitor cells that can be used to treat degenerative diseases in humans. However, there is a need to determine how ES cell-derived progenitors are regulated by both the adaptive and innate immune systems post transplantation. In this study, we demonstrate that hematopoietic progenitor cells (HPCs) derived from mouse ES cells ectopically expressing HOXB4 fail to engraft long-term in the presence of NK cells. In particular, the H60-expressing Lin(-)c-kit(+) and Lin(-)Sca-1(+) subpopulations were preferentially deleted in Rag2(-/-), but not in Rag2(-/-)gamma(c)(-/-) mice. Up-regulation of class I expression on HPCs prevented their lysis by NK cells, and Ab-mediated depletion of NK cells restored long-term HPC engraftment. In contrast to the notion that ES-derived cells are immune-privileged, we show in this study that NK cells form a formidable barrier to the long-term engraftment of ES cell-derived hematopoietic progenitors.


Asunto(s)
Antígenos de Diferenciación/biosíntesis , Citotoxicidad Inmunológica/inmunología , Células Madre Embrionarias/inmunología , Trasplante de Células Madre Hematopoyéticas , Proteínas de Homeodominio/genética , Células Asesinas Naturales/inmunología , Antígenos de Histocompatibilidad Menor/biosíntesis , Proteínas Proto-Oncogénicas c-kit/biosíntesis , Factores de Transcripción/genética , Transducción Genética , Animales , Linaje de la Célula/inmunología , Células Cultivadas , Células Madre Embrionarias/citología , Células Madre Embrionarias/metabolismo , Trasplante de Células Madre Hematopoyéticas/métodos , Proteínas de Homeodominio/inmunología , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Factores de Transcripción/inmunología
10.
J Cell Physiol ; 218(2): 256-63, 2009 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-18932230

RESUMEN

Derivation of insulin producing cells (IPCs) from embryonic stem (ES) cells provides a potentially innovative form of treatment for type 1 diabetes. Here, we discuss the current state of the art, unique challenges, and future directions on generating IPCs.


Asunto(s)
Células Madre Embrionarias/citología , Células Secretoras de Insulina/citología , Animales , Diferenciación Celular , Endodermo/citología , Humanos , Células Secretoras de Insulina/trasplante , Especificidad de Órganos , Factores de Transcripción/metabolismo
11.
J Leukoc Biol ; 83(4): 853-63, 2008 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18162511

RESUMEN

CD28 and CTLA-4 are the critical costimulatory receptors that predominantly determine the outcome of T cell stimulation, with CD28 promoting positive costimulation and CTLA-4 inducing inhibitory signals. Blockage of the B7-CD28/CTLA-4 pathway leads to transplantation tolerance. However, the exact mechanism of the inhibitory function of CTLA-4 remains elusive. Here, we investigated the influence of CTLA-4 expression on CD28 using CTLA-4-transfected Jurkat T cells as well as primary T cells. Up-regulation of CTLA-4 induced abrogation of IL-2 production, indicating an anergic phenotype of CTLA-4(high) T cells. Besides the negative signaling function of CTLA-4, we show for the first time that CTLA-4 expression promotes the down-regulation of CD28 on the T cell surface as a result of enhanced internalization and degradation of CD28. These data suggest that apart from the established competition for B7.1 and B7.2 by CTLA-4, inhibition of T cells by CTLA-4 might be additionally explained by reduction of CD28 on the cell surface, which might impede T cell response to stimulation. Our data provide a previously unrecognized mechanism for T cell regulation.


Asunto(s)
Antígenos CD/fisiología , Antígenos de Diferenciación/fisiología , Antígenos CD28/genética , Linfocitos T CD8-positivos/inmunología , Regulación de la Expresión Génica/inmunología , Linfocitos T/inmunología , Antígenos CD/genética , Antígenos de Diferenciación/genética , Antígeno CTLA-4 , Cartilla de ADN , Metabolismo Energético , Ensayo de Inmunoadsorción Enzimática , Citometría de Flujo , Humanos , Interleucina-2/análisis , Células Jurkat , Valores de Referencia , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transfección
12.
Stem Cell Res Ther ; 9(1): 204, 2018 07 27.
Artículo en Inglés | MEDLINE | ID: mdl-30053892

RESUMEN

BACKGROUND: As the prevalence of therapeutic approaches involving transplanted cells increases, so does the need to noninvasively track the cells to determine their homing patterns. Of particular interest is the fate of transplanted embryonic stem cell-derived hematopoietic progenitor cells (HPCs) used to restore the bone marrow pool following sublethal myeloablative irradiation. The early homing patterns of cell engraftment are not well understood at this time. Until now, longitudinal studies were hindered by the necessity to sacrifice several mice at various time points of study, with samples of the population of lymphoid compartments subsequently analyzed by flow cytometry or fluorescence microscopy. Thus, long-term study and serial analysis of the transplanted cells within the same animal was cumbersome, making difficult an accurate documentation of engraftment, functionality, and cell reconstitution patterns. METHODS: Here, we devised a noninvasive, nontoxic modality for tracking early HPC homing patterns in the same mice longitudinally over a period of 9 days using mesoporous silica nanoparticles (MSNs) and magnetic resonance imaging. RESULTS: This approach of potential translational importance helps to demonstrate efficient uptake of MSNs by the HPCs as well as retention of MSN labeling in vivo as the cells were traced through various organs, such as the spleen, bone marrow, and kidney. Altogether, early detection of the whereabouts and engraftment of transplanted stem cells may be important to the overall outcome. To accomplish this, there is a need for the development of new noninvasive tools. CONCLUSIONS: Our data suggest that multifunctional MSNs can label viably blood-borne HPCs and may help document the distribution and homing in the host followed by successful reconstitution.


Asunto(s)
Médula Ósea/metabolismo , Células Madre Embrionarias/metabolismo , Células Madre Hematopoyéticas/metabolismo , Nanopartículas/metabolismo , Animales , Ratones
13.
Immunol Res ; 39(1-3): 261-70, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-17917070

RESUMEN

Diabetes is a chronic autoimmune disease that affects 4-5% of the world's population. If the present trends continue, diabetes would soon become a major/leading health problem worldwide. Hence there is an urgent need to develop novel approaches for the treatment of diabetes. While transplantation of the pancreas or that of isolated pancreatic islets can lead to the cure of the disease in some patients, immunological complications and the chronic shortage of donors makes it impossible to adequately treat all patients. Interestingly, embryonic stem cells (ESC) have emerged as a possible source of pluripotent cells that can be coaxed into insulin-producing cells (IPCs) that can be used to treat diabetes. However, until appropriate protocols have been established, this new technology will be difficult to tap into. Our laboratory is interested in developing new strategies for harnessing the pluripotency of ESC and differentiating them into IPCs that are stable and will continue to produce insulin in vivo. A second aspect is the non-availability of non-invasive imaging protocols. We show here that transcriptionally targeted luciferase expression can be used successfully to non-invasively monitor the transplanted cells in vivo.


Asunto(s)
Diferenciación Celular , Células Madre Embrionarias/citología , Células Secretoras de Insulina/citología , Activinas/metabolismo , Animales , Diabetes Mellitus Tipo 1/terapia , Diagnóstico por Imagen/métodos , Células Madre Embrionarias/metabolismo , Células Madre Embrionarias/trasplante , Endodermo/citología , Endodermo/metabolismo , Humanos , Células Secretoras de Insulina/metabolismo , Células Secretoras de Insulina/trasplante , Trasplante de Islotes Pancreáticos , Luciferasas/análisis , Trasplante de Células Madre , Factores de Transcripción/metabolismo
14.
Leuk Res ; 31(2): 121-5, 2007 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-16860865

RESUMEN

The goal of this editorial is to revisit soluble human leukocyte antigens (sHLA) and to highlight the findings reported by Albitar et al. in this issue on the relation between sHLA levels in Non-Hodgkin's Lymphoma (NHL) and Hodgkin's Disease (HD). We will review key aspects of sHLA including soluble HLA-G, which has received a lot of attention in recent publications. We will then address the role of sHLA in lymphoproliferative diseases and in solid organ tumors. Lastly, we will comment on the results of Albitar et al. and their relevance to clinical application in NHL.


Asunto(s)
Antígenos HLA/inmunología , Enfermedad de Hodgkin/inmunología , Linfoma no Hodgkin/inmunología , Antígenos HLA/sangre , Enfermedad de Hodgkin/sangre , Humanos , Células Asesinas Naturales/inmunología , Linfoma no Hodgkin/sangre , Solubilidad , Linfocitos T/inmunología , Escape del Tumor/inmunología
15.
Transplantation ; 81(9): 1337-44, 2006 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-16699464

RESUMEN

BACKGROUND: T cells recognize major histocompatibility complex (MHC) molecules and their cryptic antigenic peptides on antigen-presenting cells and are generally triggered to proliferate, and when sufficient, co-stimulation is available. In soluble form, monomeric MHC molecules can induce apoptosis, anergy, or decreases of the T-cell receptor (TCR). METHODS: A dimeric fusion protein of the human leukocyte antigens (HLA)-B7 was molecularly engineered and expressed in a B-cell line to allow secretion. Alloreactive T cells were generated according to the standard protocol. RESULTS: A dimer of approximately 160 kD was obtained, affinity purified, and used to study T-cell interaction. In immobilized form, this protein efficiently stimulated alloreactive T cells to proliferate and produce interleukin (IL)-2 and interferon (IFN)-gamma in a concentration-dependent manner, up-regulating CD25 and CD69 expression. In contrast, the soluble fusion protein induced T-cell apoptosis. CONCLUSIONS: The dichotomy in T-cell regulation by a divalent MHC fusion protein warrants the use of MHC multimers as custom-designed immune-regulatory molecules both in transplantation and autoimmune disease.


Asunto(s)
Antígenos CD/inmunología , Antígenos de Diferenciación de Linfocitos T/inmunología , Antígeno B7-1/inmunología , Linfocitos T CD8-positivos/inmunología , Dipeptidil Peptidasa 4/inmunología , Antígenos HLA/inmunología , Antígenos CD/genética , Antígenos de Diferenciación de Linfocitos T/genética , Apoptosis , Linfocitos B , Línea Celular , Dimerización , Dipeptidil Peptidasa 4/genética , Humanos , Lectinas Tipo C , Proteínas Recombinantes de Fusión/inmunología , Linfocitos T Citotóxicos/inmunología , Transfección , Regulación hacia Arriba
16.
Transplantation ; 81(11): 1549-57, 2006 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-16770244

RESUMEN

BACKGROUND: Initial interruption of immunosuppression for 72 hr was analyzed in renal transplant recipients according to Calne et al.'s "window of opportunity for immunologic engagement" (WOFIE) concept. METHODS: This pilot study was designed as a randomized, open-label, prospective trial of 40 recipients (20 in the WOFIE group, 20 in the control group) of cadaveric kidney transplants who were followed up for 2 years. Immunosuppression comprised tacrolimus (trough levels 5-8 ng/mL), daclizumab (1 mg per kilogram of body weight on day 0 and after 2, 4, 6, and 8 weeks), mycophenolate mofetil (1-2 g/day), and prednisolone (maintenance dose of 10 mg/day). After induction with daclizumab, prednisolone, and mycophenolate mofetil, immunosuppression was interrupted for 72 hr in the WOFIE group. Steroid withdrawal followed in both groups within 12 to 16 weeks posttransplant. RESULTS: Patient and graft survival did not differ significantly between the two cohorts. However, the WOFIE group experienced less acute rejection episodes and developed better graft function. Although all but one of the patients in the WOFIE group successfully discontinued steroid treatment, permanent steroid withdrawal was achieved in only 76.4% of the control group. After daclizumab discontinuation, the WOFIE group demonstrated an increase of CD4CD25 T cells in peripheral blood (P<0.05 vs. control group), which was stable over time and strongly correlated with a significantly higher expression level of Foxp3-mRNA. CONCLUSIONS: Initial interruption of immunosuppression for 72 hr correlates with the induction of regulatory immunologic mechanisms and allows early and reliable minimization of immunosuppressive treatment.


Asunto(s)
Tolerancia Inmunológica/inmunología , Inmunosupresores/administración & dosificación , Trasplante de Riñón/inmunología , Linfocitos T Reguladores/inmunología , Tolerancia al Trasplante/inmunología , Adolescente , Adulto , Antígenos CD4/análisis , Esquema de Medicación , Femenino , Citometría de Flujo , Estudios de Seguimiento , Rechazo de Injerto/tratamiento farmacológico , Rechazo de Injerto/inmunología , Humanos , Tolerancia Inmunológica/efectos de los fármacos , Inmunosupresores/uso terapéutico , Riñón/inmunología , Riñón/patología , Riñón/fisiopatología , Masculino , Persona de Mediana Edad , Proyectos Piloto , Estudios Prospectivos , Receptores de Interleucina-2/análisis , Linfocitos T Reguladores/efectos de los fármacos , Factores de Tiempo , Trasplante/fisiología , Inmunología del Trasplante , Tolerancia al Trasplante/efectos de los fármacos
17.
J Leukoc Biol ; 78(3): 595-604, 2005 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-16000395

RESUMEN

Monomeric and dimeric soluble major histocompatibility complex (MHC) molecules down-regulate activated T cells in an antigen-specific manner in vitro. This property could be exploited to modulate alloresponses in vivo but has remained difficult to demonstrate. Here, intraperitoneal infusion of a Lewis-derived rat MHC class I molecule, RT1.A(l)-Fc, in Dark Agouti (RT1.A(a)) recipient rats prolonged cardiac graft survival, which led to permanent engraftment. This effect was mediated by T cell impairment of target cell lysis by CD8+ T cells and down-regulation of interferon-gamma production by CD4+ T cells. The binding of the dimeric MHC allowed ex vivo visualization of alloreactive T cells in peripheral blood, splenocytes, and allografts, revealing low frequency of alloreactive CD8+ T cells after establishment of permanent engraftment of cardiac allografts. Thus, these data show the potential of dimeric MHC molecules to promote graft survival and allow visualization of alloreactive T cells.


Asunto(s)
Rechazo de Injerto/inmunología , Supervivencia de Injerto/inmunología , Trasplante de Corazón/inmunología , Antígenos de Histocompatibilidad/administración & dosificación , Fragmentos Fc de Inmunoglobulinas/administración & dosificación , Linfocitos T/inmunología , Animales , Linfocitos T CD4-Positivos/citología , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/citología , Linfocitos T CD8-positivos/inmunología , Dimerización , Modelos Animales de Enfermedad , Supervivencia de Injerto/efectos de los fármacos , Antígenos de Histocompatibilidad/inmunología , Fragmentos Fc de Inmunoglobulinas/inmunología , Técnicas In Vitro , Infusiones Parenterales , Activación de Linfocitos/inmunología , Ratas , Ratas Endogámicas Lew , Proteínas Recombinantes de Fusión/administración & dosificación , Proteínas Recombinantes de Fusión/inmunología , Solubilidad , Linfocitos T/citología
18.
Transplantation ; 79(9): 1040-4, 2005 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-15880040

RESUMEN

BACKGROUND: Recent data show the efficacy of embryonic stem cells (ESC) to engraft in allogeneic recipients without host pretreatment. This property is due to their low expression of major histocompatibility complex (MHC) class I antigens and lack of MHC class II expression. Here, we tested the hypothesis that the constitutive FasL expression by ESC is a requirement for their stable engraftment in allogeneic recipients. METHODS: MRL and MRL-lpr/lpr mice (H-2k) were infused allogeneic 129SvJ RW-4 (H-2b) ESC without host preconditioning. The development of mixed chimerism was monitored over 100 days by flow cytometry. RESULTS: Mixed chimerism was detectable by day 7. The amount of donor cells detected varied between 3-5.5% and were lymphoid, but nonmyeloid. Only 50% of lpr/lpr mice engrafted and lost donor cells by day 28 post-ESC infusion. In contrast, >80% wild type mice engrafted and maintained mixed chimerism up to day 100. CONCLUSIONS: These data suggest a critical role for Fas-FasL engagement in ESC engraftment. We conclude that ESC may induce clonal deletion of alloreactive T cells by Fas-induced apoptosis in recipient T cells, protecting them from rejection. The data provide a rationale for improved protocols for the achievement of robust ESC-induced mixed chimerism.


Asunto(s)
Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/inmunología , Glicoproteínas de Membrana/fisiología , Trasplante de Células Madre , Quimera por Trasplante/inmunología , Receptor fas/fisiología , Animales , Línea Celular , Proteína Ligando Fas , Isoantígenos/inmunología , Ratones
19.
Transplantation ; 79(8): 897-903, 2005 Apr 27.
Artículo en Inglés | MEDLINE | ID: mdl-15849541

RESUMEN

INTRODUCTION: CTLA-4 is a negative regulatory molecule upregulated on activated T cells; however, its role in induction and maintenance of transplant tolerance is not well understood. METHODS: The characteristics and effects of a novel mouse anti-rat CTLA-4 antibody (Ab) (242B58) were examined using fluorescence-activated cell sorter, mixed lymphocyte reaction, enzyme-linked immunospot, signaling studies, and a rat model of cardiac transplant tolerance induced by administration of anti-CD28 Ab and cyclosporine. RESULTS: The anti-CTLA4 Ab was shown to bind to CTLA-4 but not prevent subsequent binding of B7 to CTLA-4. Binding to CTLA-4 did not result in phosphorylation of early cytoplasmic tyrosine kinases, suggesting that this is not a signaling Ab. However, its in vitro function was compatible with antagonization of the effects of CTLA-4, thereby increasing T-cell proliferation and interferon-gamma production in mixed lymphocyte reaction and enzyme-linked immunospot assays, respectively. Administration of 242B58 to animals treated with anti-CD28 Ab and cyclosporine either at the time of transplantation or various time-points up to 33 days posttransplantation did not result in immediate rejection, but rather caused a delayed severe acute allograft rejection at approximately 45 days posttransplant. CONCLUSIONS: Our results seem to be a reflection of the unique properties of the 242B58 Ab, which does not antagonize B7 binding to CTLA-4 and affect its ability to out-compete CD28 for B7 binding. It does, however, seem to interfere with CTLA-4 signaling, suggesting that competition for B7 may be important in induction of tolerance, but signaling through CTLA-4 is more important in maintaining a tolerant phenotype.


Asunto(s)
Antígenos de Diferenciación/metabolismo , Supervivencia de Injerto/inmunología , Trasplante de Corazón/inmunología , Tolerancia Inmunológica/inmunología , Animales , Anticuerpos/inmunología , Antígenos CD , Antígenos de Diferenciación/inmunología , Antígeno B7-1/metabolismo , Antígeno CTLA-4 , Rechazo de Injerto/inmunología , Prueba de Cultivo Mixto de Linfocitos , Ratones , Fosfotirosina/metabolismo , Unión Proteica , Proteínas Tirosina Quinasas/metabolismo , Ratas , Factores de Tiempo
20.
PLoS One ; 10(1): e0116582, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25629318

RESUMEN

Type 1 diabetes (T1D) is caused by autoimmune disease that leads to the destruction of pancreatic ß-cells. Transplantation of cadaveric pancreatic organs or pancreatic islets can restore normal physiology. However, there is a chronic shortage of cadaveric organs, limiting the treatment of the majority of patients on the pancreas transplantation waiting list. Here, we hypothesized that human iPS cells can be directly differentiated into insulin producing cells (IPCs) capable of secreting insulin. Using a series of pancreatic growth factors, we successfully generated iPS cells derived IPCs. Furthermore, to investigate the capability of these cells to secrete insulin in vivo, the differentiated cells were transplanted under the kidney capsules of diabetic immunodeficient mice. Serum glucose levels gradually declined to either normal or near normal levels over 150 days, suggesting that the IPCs were secreting insulin. In addition, using MRI, a 3D organoid appeared as a white patch on the transplanted kidneys but not on the control kidneys. These organoids showed neo-vascularization and stained positive for insulin and glucagon. All together, these data show that a pancreatic organ can be created in vivo providing evidence that iPS cells might be a novel option for the treatment of T1D.


Asunto(s)
Diferenciación Celular , Células Madre Pluripotentes Inducidas/citología , Células Secretoras de Insulina/citología , Células Secretoras de Insulina/metabolismo , Neovascularización Fisiológica , Trasplante de Células Madre , Animales , Glucemia , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Experimental/terapia , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Células Madre Pluripotentes Inducidas/ultraestructura , Imagen por Resonancia Magnética , Masculino , Ratones , Ratones Noqueados , Mitocondrias/metabolismo , Organoides , Consumo de Oxígeno
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA